You are on page 1of 32

NANOTECHNOLOGY FOR CANCER TREATMENT

FOR THE PROJECT


IN PARTIAL FULFILLMENT OF THE DEGREE
BACHELOR OF PHARMACY

SUBMITTED BY
NAME:- JATIN ISHER
ROLL NO. 1819204
B.PHARM 7 Sem

GLOBAL GROUP OF INSTITUTES, AMRITSAR


DEPARTMENT OF PHARMACY
2021

1
DECLARATION
I, Jatin Isher, hereby declare that the matter embodied in the project entitled Nanotechnology
for cancer treatment submitted to Global Group of Institutes, Amritsar (Department of
Pharmacy) is the result of my direct investigations and project has been composed under
supervision and guidance of, Mr. Mandeep Singh Chhina Assistant Professor (Department
of Pharmacy), Global Group of Institutes, Amritsar. I also declare that nothing in part or full
has been submitted for the award of any degree, diploma, or fellowship to any other Institute
or University. All the ideas and references have been duly acknowledged. If anything, wrong
is detected in future than I shall be held responsible for the same.

Date Jatin Isher

Place 1819204

Signature of Guide

2
ACKNOWLEDGEMENT

I would like to express my sincere gratitude to several individuals and organizations for
supporting me throughout my project course. I express deep sense of faith to GOD, who
graced me in finishing this project. The same perplexity haunts inside me in penning down
my sagacious sense of gratitude and admirations towards my venerable Mr. Mandeep Singh
Chhina, Assistant Professor with special thanks to Principal Dr. Manbir Kaur, Principal
for her encouragement, illuminating guidance, unflinching support, incredible perseverance
and bountiful affection sprinkled on me all the times of project. It is a matter of pleasure to
express my sense of gratitude to Ms. Gurpreet Bawa, Head of Department for their
guidance to develop independent thinking in me. I am highly thankful to my friends Aman
Vassan, Vishal Singh & Jaibir Kaur for her masterly guidance for helping inspiring help
and more over invigorate guidance, energizing criticism, constant support, and
encouragement. My family deserves special mention for their inseparable support,
unconditional love, enormous patience, unwavering support and overwhelming
encouragement, prayers and optimism which never let me in sedation during the course of
this project.

I also wish to express my sincere thanks to the Global Group of Institutes for accepting me
into the graduate program. In addition, I am deeply indebted to the department of pharmacy.

Finally, last but by no means least; also to everyone in the project, that was great sharing
premises with all of you during my project period.

Thanks for all your encouragement!

Jatin Isher

3
CONTENTS

S.No. contents Page No.


1. Introduction 6
2. Review of literature 7
2.1. Tumor targeting 8
2.2. Passive targeting via the EPR 8-9
2.3. Active targeting 9
2.4. Minimizing MPS uptake 10
2.5. NP-mediated cancer imaging 10-11
2.6. NP-mediated cancer treatments 11-12
2.7. NP-mediated drug release 12
2.8. Controlling NP-mediated drug release 12-13
2.9. Clinical candidates for NP-mediated drug delivery 13
2.10. Thermal ablative approaches to cancer treatment 13-14
2.11. Nanotechnology and photodynamic therapy 14
2.12. Nanoparticles-mediated gene therapy 14-15
2.13. Multimodality NPs for cancer treatment 15-16
2.14. NP-related toxicity 16
3. Conclusion 17-18
4. References 19-32

4
CHAPTER-1
INTRODUCTION

5
CHAPTER 1

INTRODUCTION

The need for an advanced technology that will play a key role in cancer treatment is clearly
evident in statistics showing that cancer incidence, prevalence, and mortality remain at an all-
time high [1]. Cancer is one of the leading causes of death worldwide as an estimated 7.6
million people die each year and make up 13% of all deaths. The cancer-related death rate is
expected to rise to 13.1 million by 2030. Cancer is not a single disease but a host of diseases
per organ or system that develops a set of different diseases. Most cases of cancer can be
avoided, with some estimates showing that about 30 percent of cancer deaths are associated
with smoking or other lifestyle factors or eating habits that may be avoided by changes in
human behavior [2-4]. However, most cancers are unavoidable with simple behavioral
changes and require the invention of technology to improve outcomes. The developed world
has had significant successes in reducing cancer-causing cancers [e.g., human papilloma virus
(HPV)] [5-7]. This success can be enhanced by the widespread use of existing vaccine
technology as well as the use of nanotechnology and other technologies to improve vaccine
efficacy [5-8]. Nanotechnology may also be able to increase the percentage of early detection
cancers through improved imaging and this, in line with the aggressive implementation of
existing screening technologies, will lead to improved outcomes for cancer patients [9-10].
However, for many types of cancer, new therapies for established diseases are needed. To
address these medical needs, nano-sized cellular devices capable of distinguishing between
harmful and non-harmful cells and delivering lethal load must be developed. This review
summarizes many of the new technologies that have been reported in recent years and that
promise to improve outcomes for cancer patients.

6
CHAPTER-2
REVIEW OF
LITERATURE

7
CHAPTER 2

REVIEW OF LITERATURE

2.1. Tumor targeting

One of the most important potential benefits of nanotechnology in the treatment of cancer is
to identify the tumor. The ability to distinguish lethal cells from non-malignant cells and to
eliminate harmful cells by choice is fundamental to the work of nanotechnology as it is
associated with cancer treatment. Two important processes are involved in distinguishing
between harmful and malignant cells: active and passive targeting. Passive targeting takes
benefits of the enhanced permeability and retention (EPR) effect [11-12] to increase the
saturation of nanoparticles (NPs) in the plant. Active identification [13] may involve the
identification of selective antigen-presenting cells, usually proteins expressed in areas above
the cancer cells to incorporate NPs into malignant cells or, alternatively, exploit biochemical
structures associated with malignancies such as matrix metalloproteinase secretion [14].
Active and passive targeting can be used independently, or the two approaches can be
combined. Both strategies benefit from the local modification of NPs to reduce the
acquisition of the macrophage phagocytic system (MPS) [15], thus, increasing the cycle time.

2.2. Passive targeting via the EPR

It is well known that tumor vasculature leaks when it is related to the hierarchical structure of
normal vasculature, in part, because killer cells do not respond to cell expression required for
systematic vasculogenesis [16-17]. Macromolecules can enter the tumor through a leaky
vasculature and develop, in part, due to a decrease in lymph clearance [18] in tumors with a
substance called enhanced permeability and end-of-life (EPR) effect [19]. The effectiveness
of EPR depends on the size of the tumor, the type of tumor, and the tumor heterogeneity,
among other factors. The effectiveness of EPR also depends largely on the size of the
targeted treatment. As explained by Maeda, EPR-based localization works by operating over
a range of MW 40 kDa – 800 kDa, which in turn around the earth is accompanied by a radius
of 2.3 to 6.1 nm [20]. Preferred localization of protein synthesis in plant tissues by EPR has
surprisingly revealed a minimum of 25 kDa with improved intake of large or small proteins -
although small peptides required active guidance to be maintained. In contrast, liposomes did
not benefit from functional guidance [20-21]. A variety of NPs have been shown to be
present in plant genes through EPR including multiwalled carbon nanotube (MWNT), single-

8
walled carbon nanotube (SWNT) [22-23], and liposomes [24], as well as infected NPs [25].
NPs may vary greatly in density, and some features from global proteins and several large
NPs of nanometres in one area have been reported to be localized in plant tumor with EPR.
To our knowledge, the efficacy associated with local implantation with EPR in various NPs
has not been systematically investigated in any implant model. An interesting variation of
EPR-indirect regulation has recently been described in which gold nanorods are introduced
into the tumor tissue via EPR and are used to heat the tumor in laser light. The procedure was
followed by the introduction of the anti-cancer agent ADHGM into cancer cells in
recognition of GRP78 which was regulated in prostate cancer cells in response to increasing
temperature [26].

2.3. Active targeting

In fact, any ligand that exhibits a special malignant binding in relation to non-malignant cells
or leads to selective opening close to malignant cells [27, 28] can be used to identify
malignant cells. In this regard, receptor factor receptors such as epidermal growth factor
receptor (EGFR) [29], transferrin [30-33], death receptor (DR) complexes (e.g., DR5 [34, 35]
), and folate ligand [36-40] and tumor-specific antigens (e.g., PSMA [41- 42]) have all been
used to make local NPs in malignant cells with active targeting. A variety of chemical and
biological agents have been used to target NPs in malignant cells that express molecular
target receptors including monoclonal antibodies [43], small molecules, and nucleic acid
aptamers [44-45]. Factors contributing to a specific type of targeted molecule specifically
used include molecular weight (MW), targeted correlation, valency, and biocompatibility.
Although effective identification is conceptually straightforward, this type of identification
does not in the same way improve the local performance of a plant. For example, monoclonal
antibody (mAb) administration was found in some cases to not improve local tumor
production [46]. In addition, effective direction may influence other variables, such as the
cycle, and these indirect effects may confuse direct identification results. Using variable
ligand values of the target ligand, it was shown that the active direction of NPs affects
cellular detection within the tumor, but not the direction of the tumor itself [30]. Thus,
effective identification remains an important strategy for local NP; however, caution should
be used in interpreting the observed biological effects in active guidance.

9
2.4. Minimizing MPS uptake

The accumulation of NPs in tumor tissue requires a longer period of circulation and
avoidance by retaining the reticuloendothelial system (RES) (aka MPS [47]). The
incorporation of NPs by polyethylene glycol (PEG) or other amphipathic agents reduces the
binding of proteins involved in the activation of NPs and, thus, reduces the detection of MPS
[48]. PEGylation reduced MPS detection of quantum dots (QDs) by up to nine times, whereas
peptide release had a small effect [49]. Dai and colleagues showed that by using 90 kDa
amphiphilic poly (maleic anhydride-alt-1-octadecene) -methoxy poly (ethylene glycol) [C18-
PMH-mPEG], they were able to obtain 30% of the controlled SWNT dose. Modified on
tumor tissue [22-23]. Recent studies have examined the effects of local modification of gold
nanoparticles (GNPs) on interactions with blood components including NP bio-distribution
[50]. GNPs are internalized by monocytes regardless of spatial modification. Improved plant
collection was associated with improved circulation and was found to be more dependent on
new PEG, than lyophilized, which improves over time. The effects of overcharging on cell
capture and distribution of PEGoligocholic acid micelles were systematically evaluated [51].
Low negative charge charges have been found to increase tumor appearance and reduce
absorption of MPS cells in the liver. Landscaping to reduce MPS acquisition continues to be
an important strategy for developing NPs through improved medical care. For example, low-
dose MW has been developed as an alternative to PEGylation that may allow for the
maintenance of certain cell-secreted secretions by PEG [52].

2.5. NP-mediated cancer imaging

NPs may be particularly useful in image processing [53] due to the high localization to
volume (relative to large particles) and the potential for multiple chemical modification sites
that can be used to enhance image sensitivity [53]. Although avoiding taking macrophage is
important for NP-mediated effects in most cases, the tendency of NPs to enter macrophage-
mediated phagocytosis may be beneficial in image processing. Super paramagnetic iron oxide
NPs (IONPs) have been used to capture MR of lymph nodes following macrophage
absorption, which may be beneficial in detecting metastatic disease [54-55]. Negative
lymphatic flow of tumors that contributes to the accumulation of NPs for drug delivery may
be used to capture tumors with IONPs [56]. IONPs were also synthesized with an amino-
terminal component of the urokinase plasminogen activator to mimic breast cancer [57],
whereas antibody binding to EGFR was used to target brain tumors [58]. Common chemical

10
mechanisms for the development of IONP modified topical cancer imaging are improved
[59]. Recently, a new in vivo combination of NPs with imaging agents has been described
[60].

Several different types of NP have been synthesized with paramagnetic Gd3 + chelates to
improve MR differentiation including dendrimers, micelles, and cNTs [61]. In fact, very clear
photography of small numbers of harmful cells can be achieved by combining targeted
agents, such as mAb, and Gd3 + -chelates to affect MR relaxation or interaction with other
imaging probes. In fact, sensitivity is a major problem in photographic research. Another
possible way is to amplify the signal in an area of interest by delivering the right enzyme. For
example, horseradish peroxidase has been introduced into xenograft tumors by binding to
tumor-specific mAb, and this has been used to oligomerize MRspecific ligands to achieve
improved signal detection and imaging [62]. GNPs have also been used to enhance contrast
in x ray images that provide triiodobenzene-related benefits [63]. In addition to enhancing
image imaging comparisons, GNPs affect X-ray scatter and can be used to generate local
radiation and improve therapeutic effects [64-65].

In fact, NPs can be used for both imaging and treatment applications [66]. For example, TiO2
NPs may be used both to enhance CT imaging (computed tomography) and as a
photodynamic therapy sensor [67]. Magnetic NP can be used for both MR imaging
enhancement and hyperthermia applications for advanced cancer treatment [68]. The αvβ3
integrin-specific peptide motif RGD may be used to target IONPs in malignant cells in both
acute and chronic hyperthermia [69]. IONPs can be combined with methotrexate [70],
paclitaxel (PTX) [71], or other anti-cancer drugs [72] through theranostic (therapeutic +
diagnostic) applications. Gold NPs, quantum dots, and cNTs have also been modified and
used for potential theranostic applications [69].

2.6. NP-mediated cancer treatments

NPs used, or developed, for cancer treatment are usually not naturally cytotoxic. Therefore,
NPs must modify the chemical and / or physical environment especially in the region closest
to the cancer cell in order to use cytotoxicity. As mentioned in previous sections, NPs
targeted harmful cells mainly by inactive EPR targeting and / or active targeting, usually
based on specific cell recognition events such as EGF / EGFR interactions. Once implanted
in a tumor, NPs trigger a cytotoxic response to cancer cells usually using one of three
methods: (1) drug release [73], (2) hyperthermia or thermal discharge [74], and (3) reactive

11
oxygen species (ROS)) - mediated killing [75]. These methods can be used independently or
can be used together in the form of multimodality in the treatment of cancer. The advantages
associated with non-NP-mediated withdrawal methods are that NPs may mediate local effects
based on cellular recognition events at cellular level. Thus, by directing NPs to specific cells
(e.g., malignant cells), one can improve neoplastic tissue elimination while limiting damage
to normal or nearby normal cells. This type of procedure is especially important in highly
invasive diseases, such as glioblastoma multiforme (GBM), in which the malignant cells
cannot be differentiated by standing in non-malignant cells.

2.7. NP-mediated drug release

NP-directed drug delivery is based on the premise that, for the most part, it is less difficult to
kill a cancer cell than any other non-sick cell. Ordinary cytotoxic agents, such as doxorubicin
(DOX), are highly cytotoxic to cancer cells but, unfortunately, are highly cytotoxic to
harmless cells as well - especially cells that quickly divide into the intestinal tract and bone
marrow. NP-mediated delivery of common cytotoxic drugs allows control over drug
cytotoxicity based on NP bio distribution profile instead of free drug [76-77]. NP-directed
drug delivery also reduces the rate of low MW cytotoxic discharge which provides a growing
opportunity to remain distributed and accumulated in the target area. A successful example of
nanotechnology-led drug delivery is the liposome-mediated delivery of DOX (e.g., Doxil)
[78] which significantly reduced cardiac toxicity [79] in relation to free DOX. The albumin-
conjugated PTX NP (Abraxane) has shown promising efficacy in breast cancer and ovarian
cancer and has been approved by the FDA [80-81]. The field of nanotechnology addressed
the problem related to the hydrophobicity of PTX and assisted in the preparation of free-form
solvent (cremphor) -free that reduced total therapeutic toxicity [80]. Many recent studies have
also suggested new approaches to improved drug delivery using NPs. Our laboratory has
shown that the creation of a nano-size DNA polymer results in improved antileukemic
activity associated with low MW drugs [82].

2.8. Controlling NP-mediated drug release

The use of NPs in drug delivery requires the release of the drug from the tumor site or to
dangerous cells when implanted internally. Therefore, strategies to improve drug release in
the plant environment are an integral part of therapeutic NP-based therapeutic strategies. One
of the potential problems with current drug delivery methods is that the drug is only gradually
released from the NP following local to-plant tissue production with EPR. This slow release

12
may result in lower levels of free drugs that are insufficient to use biological, e.g., cytotoxic
response [73]. Hot labeled liposomes are developed to accelerate drug delivery following
localization of the tumor with EPR, a technique that advances in clinical trials [73].

2.9. Clinical candidates for NP-mediated drug delivery

The key factors in the effective use of NPs as therapeutic agents that include effective and
efficient identification and prevention of MPS have been developed to the point that people
using NP-based therapy without liposomes undergo clinical trials and exhibit drug
withdrawal and toxicity profiles. Significant improvements related to conventional
chemotherapy. A specific NP event that combines EPR activity regulation with active
targeting and immune cell proliferation is BIND-014, which has recently entered clinical
trials [41]. BIND-014 uses RNA aptamer A10-03 to locate NP in prostate cancer cells and
release docetaxel chemotherapy. PEG is used to reduce NP detection by MPS. In addition to
NP use in drug delivery, psychiatric NPs also undergo clinical trials such as [18F] -FAC
family of PETimaging agents tested for dosage of therapeutic agents such as gemcitabine,
cytarabine, and fludarabine [83-84]. Another highly promising example is NPs containing
different magnetic resonance imaging (MRI) agents targeting αvβ3- intregrin found on the
surface of newly developed blood vessels associated with early tumor growth [85-86]. The
viral nanoparticle (VPN) is designed for genetic treatment against leukemia [87].
Cyclodextrin-based NP that safely binds a small RNA (siRNA) agent that is able to shut
down an important enzyme in cancer cells is also under clinical examination [88-90].
Cyclodextrin-conjugated camptothecin polymeric NP is currently under clinical trials [91]. In
a joint study between Harvard Medical School and MIT, clinical research is undergoing a
process to determine the novel class of super paramagnetic NPs to identify premetastatic
circulating cells [92].

2.10. Thermal ablative approaches to cancer treatment

Local ablative methods [93] including radiofrequency ablation (RFA) [94-95], laserinduced
thermotherapy (LITT) [96], and microwave ablation [97] are widely used in the treatment of
metastatic disease [98], especially the lungs [99] and the liver [100] in various large plants.
These thermal methods currently used do not use NPs; therefore, the implementation is based
on the detection of macroscopic metastatic lesions instead of the recognition of specific
molecules as is possible with the use of NP mediation interventions. Radiologic guidance can
improve tumor specificity [101-102] and increase the effectiveness of cancer treatment. It is

13
important to note that micrometastatic disease, especially in areas far from a large plant, is a
very bad sign of prediction. Therefore, the use of nanotechnology methods to eliminate
micrometastatic disease represents one of the most important purposes of using
nanotechnology in the treatment of cancer. Although not yet clear at the cellular level, current
therapies have a high success rate with a positive response of up to 97% using RFA and 98%
using LITT to treat breast metastases [103] and colorectal cancer metastases [104].
Cryoablation uses low local temperatures to freeze and kill neoplastic tissue [105]. Our
laboratory has shown that MNTs implanted in DNA have the potential to eradicate prostate
cancer [74] in the model of naked mice in infrared radiation (NIR), without causing
significant damage to nearby tissues. DNA enhanced the dispersion of MWNTs, like other
NTs, stimulating NIR radiation into the muscle. The use of double-stranded DNA (dsDNA) -
complex SWNTs in combination with mAb for selective extraction and thermal extraction
has recently been described [106-107] as a possible cancer treatment. Focal ablative therapy
is considered an alternative form of surgery and radiation therapy for prostate cancer [108]. It
is important to note that in addition to any direct thermal effect linked to NPs, ablative
treatment also modulates the immune response, and this will affect the overall antitumor
response [109].

2.11. Nanotechnology and photodynamic therapy

The cytotoxic effects of photosensitizing porphyrins in conjunction with exposure to light


have been well documented, and photodynamic therapy (PDT) is widely used to treat bladder
cancer [110], esophageal cancer [111], and other fatal diseases and other neoplastic
conditions such as degeneration [112]. NP-mediated deployment of photosensitizing
porphyrins [113] is expected to provide several similar benefits associated with the delivery
of NP-induced cytotoxic drugs including intra-intratumoral local stimulation caused by
enhanced penetration and retention (EPR) effect [12] and toxicity reduction. Become
systemic and, in the case of photoactive compounds, reduce light sensitivity [114].
Alternatively, locally controlled NPs are built, in part; Photosensitizing porphyrins are
expected to be stored in a targeted tissue that allows more exposure to light by a single
controlled dose.

2.12. Nanoparticles-mediated gene therapy

Although it has been known for decades that DNA is the basis for life-sustaining cells that
carry information from one generation to the next, until nanotechnology begins to use DNA

14
to make macromolecules or to produce biochips, some of these bio molecule’s functions have
not yet been accomplished. The role of cellular DNA is limited, perhaps, because of the
limitations imposed on the structure and binding between the corresponding strands. In
addition to these cellular roles, nanotechnology is now discovering much of the hidden power
of DNA. By exploiting its amphipathic properties, single-stranded DNA (ssDNA) sequences
can be used to decompose hydrophobic NPs such as carbon nanotubes (cNTs) to make them
suitable for vivo use. DNA sequences have the ability to process information in biological
experiments. Its structure and compact properties have made it ideal for planning NP
planning in biochip and biosensor production.

Antisense gene therapy is a potentially powerful tool for both biomedical research and
treatment of various diseases, including cancer. Although the potential for antisense gene
therapy has been recognized for decades, its advances in effective treatment have met with
challenges in terms of viral transmission, DNAse degradation, cellular invasion, and viral
toxicity. Over the past few years, several researchers have demonstrated the potential for
increased gene therapy with the help of nanotechnology [87, 88, 90, 115, and 116]
addressing the prevalence of these problems and successfully interpreted them in clinical
trials.

2.13. Multimodality NPs for cancer treatment

Abscesses come in many forms, including many cell types and complex interactions between
cell components that contribute to treatment. One of the potential benefits of nanotechnology
is the ability to deliver and / or apply more than one therapeutic approach. Our laboratory is
investigating a multimodality NP novel that has shown strong antitumor activity through
simple ROS generation through DNA extraction. An example of nanomaterials that have
been used for multimodality applications, including drug delivery and thermal extraction,
cNTs [117]. Heat alone cannot possibly be a good way to cause the death of a plant cell as
heat can be eliminated by blood vessels. Heat may also improve chemotherapeutic efficacy
(hyperthermia), and heat-induced chemotherapy releases may increase or improve direct heat
output. Malnutrition, hypoxia, and acidic pH have all been shown to alert tumor cells to
hyperthermia [118-119].

Chemosensitization caused by hyperthermia can improve the effectiveness of chemotherapy,


and multi-modality NPs can be used in both drug delivery and the promotion of
hyperthermia. Part of the hormone FGF was used to target gold NPs in U2OS cancer cells

15
that were transmitted by fibroblast growth factor receptor (FGFR), and it was shown that cell
death could be caused by exposure to NIR irradiation [120]. Gold nanospheres have recently
been loaded with DOX and targeted to EphB4-producing plants using a peptide identified
using a phage signal [121]. Hyperthermia was promoted by NIR burns; however, other
mechanisms involving the use of flexible magnets in conjunction with IONPs [122] are
explored in pre- clinical and clinical studies, as well as in the use of high-intensity focused
ultrasound (HIFU) hyperthermia in combination with thermo-sensitive liposomes (TSL).
Recently described [123]. Drug release caused by temperature in liposomes is expected to
improve local drug concentration in therapeutic performance that improves the tumor without
increasing systemic toxicity. Although temperatures above 42 ° C may block blood flow,
temperatures above 41–42 ° C can significantly improve the effects of chemotherapy and
radiation. For example, 30% less radiation is needed to kill cells heated to 42 ° C relative to
body temperature [124], although technically it is difficult to maintain the temperature of the
tissue in a controlled manner above body temperature. MR is important in drawing
temperature, and MR-HIFU systems can be used to control local temperatures.

2.14. NP-related toxicity

One of the potential dangers of using nanomaterials in cancer treatment and human health is,
in general, the potential for toxicity [68, 75, and 125]. Nanomaterials are different in
chemical composition, charging, and even a specific size, therefore, standard toxicity
statements are almost impossible. Another major concern is that NPs may be carcinogenic,
for example, causing an increase in ROS production and leading to DNA mutations. NP
exposure is associated with asthma, respiratory tract infections, Alzheimer's disease, and
Parkinson's disease. Various vascular-related events such as blood clots are associated with
NPs entering the circulatory system. Further research is needed to determine the actual risks
associated with NP use and to determine the potential benefits outweigh the risks.

16
CHAPTER-3
DISSCUSSION &
CONCLUSION

17
CHAPTER-3

DISCUSSION &CONCLUSION

Nanotechnology plays a very important role in the diagnosis and treatment of cancer. The
size regime of NPs is relatively small compared to cells and cellular organelles that allow
NPs to interact with specific cellular features and allow the localization of tumor cells by
active targeting . The size regime of NPs is also appropriate for passive targeting to tumor
tissue via the EPR. Therefore, nano-sized materials have certain cancer treatment benefits
with distinct drug-related properties. These properties are used successfully in advanced
delivery of chemotherapeutic drugs leading to improved cancer-fighting function and
reduction of systemic toxicity.

The chemical diversity of NPs allows interactions with magnetic fields, NIR irradiation, and
other external fields to provide a more precise interaction between the outer fields of plant
tissue and potentially harmful individual cells in vivo. The versatility of NPs also allows for
external field distortion that provides improved visual acuity of image applications. The
unparalleled specificity of the link between the outer fields and the malignant cells in the
normal tissue context provided by appropriate NPs is expected to lead to more accurate and
earlier diagnosis and improved treatment outcomes. Another concern that may limit the
effectiveness of other NPs in the treatment of cancer is the toxicity of nanomaterials that
require further investigation. However, advanced cancer treatments using nanotechnology
will continue to be developed and result in improved treatment outcomes.

18
CHAPTER-4
REFERENCES

19
CHAPTER-4

REFERENCES

1. Ferlay J, Shin HR, Bray F, Forman D, Mathers C, Parkin DM. Estimates of worldwide
burden of cancer in 2008: GLOBOCAN 2008. Int. J. Cancer. 2010; 127:2893–2917.
[PubMed: 21351269]

2. Barnard RJ. Prevention of cancer through lifestyle changes. Evid. Based Complementary
Altern. Med. 2004; 1:233–239.

3. Holmes RS, Vaughan TL. Epidemiology and pathogenesis of esophageal cancer. Semin.
Radiat. Oncol. 2007; 17:2–9. [PubMed: 17185192]

4. Watabe K, Nishi M, Miyake H, Hirata K. Lifestyle and gastric cancer: a case-control


study. Oncol. Rep. 1998; 5:1191–1194. [PubMed: 9683833]

5. Moniz M, Ling M, Hung CF, Wu TC. HPV DNA vaccines. Front. Biosci. 2003; 8:d55–
d68. [PubMed: 12456324]

6. Parkin DM, Bray F. Chapter 2: The burden of HPV-related cancers. Vaccine. 2006;
24(Suppl 3) S3/11–25.

7. Walboomers JM, Jacobs MV, Manos MM, Bosch FX, Kummer JA, Shah KV, Snijders PJ,
Peto J, Meijer CJ, Munoz N. Human papillomavirus is a necessary cause of invasive cervical
cancer worldwide. J. Pathol. 1999; 189:12–19. [PubMed: 10451482]

8. Paavonen J, Naud P, Salmeron J, Wheeler CM, Chow SN, Apter D, Kitchener H,


Castellsague X, Teixeira JC, Skinner SR, Hedrick J, Jaisamrarn U, Limson G, Garland S,
Szarewski A, Romanowski B, Aoki FY, Schwarz TF, Poppe WA, Bosch FX, Jenkins D,
Hardt K, Zahaf T, Descamps D, Struyf F, Lehtinen M, Dubin G. Efficacy of human
papillomavirus (HPV)-16/18 AS04-adjuvanted vaccine against cervical infection and
precancer caused by oncogenic HPV types (PATRICIA): final analysis of a double-blind,
randomised study in young women. Lancet. 2009; 374:301–314. [PubMed: 19586656]

9. Nystrom L, Rutqvist LE, Wall S, Lindgren A, Lindqvist M, Ryden S, Andersson I,


Bjurstam N, Fagerberg G, Frisell J, Tabar L, Larsson L-G. Breast cancer screening with
mammography: overview of Swedish randomised trials. Lancet. 1993; 341:973–978.
[PubMed: 8096941]

20
10. Winawer S, Fletcher R, Rex D, Bond J, Burt R, Ferrucci J, Ganiats T, Levin T, Woolf S,
Johnson D, Kirk L, Litin S, Simmang C. Colorectal cancer screening and surveillance:
clinical guidelines and rationale-Update based on new evidence. Gastroenterology. 2003;
124:544–560. [PubMed: 12557158]

11. Maeda H. The enhanced permeability and retention (EPR) effect in tumor vasculature: the
key role of tumor-selective macromo-lecular drug targeting. Adv. Enzyme Regul. 2001;
41:189–207. [PubMed: 11384745]

12. Maeda H. Macromolecular therapeutics in cancer treatment: the EPR effect and beyond.
J. Control. Release. 2012; 164:138–144. [PubMed: 22595146]

13. Byrne JD, Betancourt T, Brannon-Peppas L. Active targeting schemes for nanoparticle
systems in cancer therapeutics. Adv. Drug Deliv. Rev. 2008; 60:1615–1626. [PubMed:
18840489]

14. Hongmei, Li SSY.; Miteva, M.; Nelson, CE.; Werfel, T.; Giorgio, TD.; Duvall, CL.
Matrix metalloproteinase responsive, proximity-activated polymeric nanoparticles for siRNA
delivery. Adv. Funct. Mater. 2013; 23:3040–3052. [PubMed: 25214828]

15. Nie S. Understanding and overcoming major barriers in cancer nanomedicine.


Nanomedicine (Lond). 2010; 5:523–528. [PubMed: 20528447]

16. Shchors K, Evan G. Tumor angiogenesis: cause or consequence of cancer? Cancer Res.
2007; 67:7059–7061. [PubMed: 17671171]

17. Weis SM, Cheresh DA. Tumor angiogenesis: molecular pathways and therapeutic targets.
Nat. Med. 2011; 17:1359–1370. [PubMed: 22064426]

18. Yu MK, Park J, Jon S. Targeting strategies for multifunctional nanoparticles in cancer
imaging and therapy. Theranostics. 2012; 2:3–44. [PubMed: 22272217]

19. Maeda H. Tumor-selective delivery of macromolecular drugs via the EPR effect:
background and future prospects. Bioconjug. Chem. 2010; 21:797–802. [PubMed: 20397686]
20. Schmidt MM, Wittrup KD. A modeling analysis of the effects of molecular size and
binding affinity on tumor targeting. Mol. Cancer Ther. 2009; 8:2861–2871. [PubMed:
19825804]

21
21. Wittrup KD, Thurber GM, Schmidt MM, Rhoden JJ. Practical theoretic guidance for the
design of tumor-targeting agents. Method Enzymol: Protein Engineering for Therapeutics
Vol203, Pt B. 2012; 503:255–268.

22. Liu Z, Chen K, Davis C, Sherlock S, Cao Q, Chen X, Dai H. Drug delivery with carbon
nanotubes for in vivo cancer treatment. Cancer Res. 2008; 68:6652–6660. [PubMed:
18701489]

23. Robinson JT, Hong G, Liang Y, Zhang B, Yaghi OK, Dai H. In vivo fluorescence
imaging in the second near-infrared window with long circulating carbon nanotubes capable
of ultrahigh tumor uptake. J. Am. Chem. Soc. 2012; 134:10664–10669. [PubMed: 22667448]

24. Maruyama K. Intracellular targeting delivery of liposomal drugs to solid tumors based on
EPR effects. Adv. Drug Deliv. Rev. 2011; 63:161–169. [PubMed: 20869415]

25. Wen AM, Shukla S, Saxena P, Aljabali AA, Yildiz I, Dey S, Mealy JE, Yang AC, Evans
DJ, Lomonossoff GP, Steinmetz NF. Interior engineering of a viral nanoparticle and its tumor
homing properties. Biomacromolecules. 2012; 13:3990–4001. [PubMed: 23121655]

26. Gormley AJ, Larson N, Sadekar S, Robinson R, Ray A, Ghandehari H. Guided delivery
of polymer therapeutics using plasmonic photothermal therapy. Nano Today. 2012; 7:158–
167. [PubMed: 22737178]

27. Danhier F, Feron O, Preat V. To exploit the tumor microenvironment: passive and active
tumor targeting of nanocarriers for anti-cancer drug delivery. J. Control. Release. 2010;
148:135–146. [PubMed: 20797419]

28. Mail JL, Raki M, Bergström KA, Uutela P, Lehtinen K, Hiltunen A, Pikkarainen J, Liang
H, Pitkänen S, Määttä A-M, Ketola RA, Yliperttula M, Wirth T, Urtti A. Pre-targeting and
direct immunotargeting of liposomal drug carriers to ovarian carcinoma. PLoS One. 2012;
7:e41410. [PubMed: 22844475]

29. Yang L, Mao H, Wang YA, Cao Z, Peng X, Wang X, Duan H, Ni C, Yuan Q, Adams G,
Smith MQ, Wood WC, Gao X, Nie S. Single chain epidermal growth factor receptor antibody
conjugated nanoparticles for in vivo tumor targeting and imaging. Small. 2009; 5:235–243.
[PubMed: 19089838]

22
30. Choi CH, Alabi CA, Webster P, Davis ME. Mechanism of active targeting in solid tumors
with transferrin-containing gold nanoparticles. Proc. Natl. Acad. Sci. USA. 2010; 107:1235–
1240. [PubMed: 20080552]

31. Huang RK, Steinmetz NF, Fu CY, Manchester M, Johnson JE. Transferrin-mediated
targeting of bacteriophage HK97 nanoparticles into tumor cells. Nanomedicine (Lond.).
2011; 6:55–68. [PubMed: 21182418]

32. Ishida O, Maruyama K. Transferrin conjugated PEG-liposomes as intracellular targeting


carrier for tumor therapy. Nihon Rinsho. 1998; 56:657–662. [PubMed: 9549352]

33. Li JL, Wang L, Liu XY, Zhang ZP, Guo HC, Liu WM, Tang SH. In vitro cancer cell
imaging and therapy using transferrin-conjugated gold nanoparticles. Cancer Lett. 2009;
274:319–326. [PubMed: 18977071]

34. Fay F, McLaughlin KM, Small DM, Fennell DA, Johnston PG, Longley DB, Scott CJ.
Conatumumab (AMG 655) coated nanoparticles for targeted pro-apoptotic drug delivery.
Biomaterials. 2011; 32:8645–8653. [PubMed: 21875750]

35. Ding B, Wu X, Fan W, Wu Z, Gao J, Zhang W, Ma L, Xiang W, Zhu Q, Liu J, Ding X,


Gao S. Anti-DR5 monoclonal antibody-mediated DTIC-loaded nanoparticles combining
chemotherapy and immunotherapy for malignant melanoma: target formulation development
and in vitro anticancer activity. Int. J. Nanomed. 2011; 6:1991–2005.

36. Dixit V, Van den Bossche J, Sherman DM, Thompson DH, Andres RP. Synthesis and
grafting of thioctic acid-PEG-folate conjugates onto Au nanoparticles for selective targeting
of folate receptor-positive tumor cells. Bioconjug. Chem. 2006; 17:603–609. [PubMed:
16704197]

37. Esmaeili F, Ghahremani MH, Ostad SN, Atyabi F, Seyedabadi M, Malekshahi MR,
Amini M, Dinarvand R. Folate-receptor-targeted delivery of docetaxel nanoparticles prepared
by PLGAPEG-folate conjugate. J. Drug Target. 2008; 16:415–423. [PubMed: 18569286]

38. Kularatne SA, Low PS. Targeting of nanoparticles: folate receptor. Methods Mol. Biol.
2010; 624:249–265. [PubMed: 20217601]

39. Schroeder JE, Shweky I, Shmeeda H, Banin U, Gabizon A. Folate-mediated tumor cell
uptake of quantum dots entrapped in lipid nanoparticles. J. Control. Release. 2007; 124:28–
34. [PubMed: 17928088]

23
40. Song EQ, Zhang ZL, Luo QY, Lu W, Shi YB, Pang DW. Tumor cell targeting using
folateconjugated fluorescent quantum dots and receptor-mediated endocytosis. Clin. Chem.
2009; 55:955–963. [PubMed: 19282359]

41. Hrkach J, Von Hoff D, Mukkaram Ali M, Andrianova E, Auer J, Campbell T, De Witt D,
Figa M, Figueiredo M, Horhota A, Low S, McDonnell K, Peeke E, Retnarajan B, Sabnis A,
Schnipper E, Song JJ, Song YH, Summa J, Tompsett D, Troiano G, Van Geen Hoven T,
Wright J, LoRusso P, Kantoff PW, Bander NH, Sweeney C, Farokhzad OC, Langer R, Zale
S. Preclinical development and clinical translation of a PSMA-targeted docetaxel
nanoparticle with a differentiated pharmacological profile. Sci. Transl. Med. 2012; 4 128ra39.

42. Kasten BB, Liu T, Nedrow-Byers JR, Benny PD, Berkman CE. Targeting prostate cancer
cells with PSMA inhibitor-guided gold nanoparticles. Bioorg. Med. Chem. Lett. 2013;
23:565–568. [PubMed: 23232055]

43. El-Sayed IH, Huang X, El-Sayed MA. Selective laser photothermal therapy of epithelial
carcinoma using anti-EGFR antibody conjugated gold nanoparticles. Cancer Lett. 2006;
239:129–35. [PubMed: 16198049]

44. Farokhzad OC, Cheng J, Teply BA, Sherifi I, Jon S, Kantoff PW, Richie JP, Langer R.
Targeted nanoparticle-aptamer bioconjugates for cancer chemotherapy in vivo. Proc. Natl.
Acad. Sci. USA. 2006; 103:6315–6320. [PubMed: 16606824]

45. Yu C, Hu Y, Duan J, Yuan W, Wang C, Xu H, Yang XD. Novel aptamer-nanoparticle


bioconjugates enhances delivery of anticancer drug to MUC1-positive cancer cells in vitro.
PLoS One. 2011; 6:e24077. [PubMed: 21912664]

46. Singh R, Erickson HK. Antibody-cytotoxic agent conjugates: preparation and


characterization. Methods Mol. Biol. 2009; 525:445–467. xiv. [PubMed: 19252846]

47. Jokerst JV, Lobovkina T, Zare RN, Gambhir SS. Nanoparticle PEGylation for imaging
and therapy. Nanomedicine (Lond.). 2011; 6:715–728. [PubMed: 21718180]

48. Romberg B, Hennink WE, Storm G. Sheddable coatings for long-circulating


nanoparticles. Pharm. Res. 2008; 25:55–71. [PubMed: 17551809]

49. Schipper ML, Iyer G, Koh AL, Cheng Z, Ebenstein Y, Aharoni A, Keren S, Bentolila LA,
Li J, Rao J, Chen X, Banin U, Wu AM, Sinclair R, Weiss S, Gambhir SS. Particle size,

24
surface coating, and PEGylation influence the biodistribution of quantum dots in living mice.
Small. 2009; 5:126– 134. [PubMed: 19051182]

50. Shah NB, Vercellotti GM, White JG, Fegan A, Wagner CR, Bischof JC. Blood-
nanoparticle interactions and in vivo biodistribution: impact of surface PEG and ligand
properties. Mol. Pharm. 2012; 9:2146–2155. [PubMed: 22668197]

51. Xiao K, Li Y, Luo J, Lee JS, Xiao W, Gonik AM, Agarwal RG, Lam KS. The effect of
surface charge on in vivo biodistribution of PEG-oligocholic acid based micellar
nanoparticles. Biomaterials. 2011; 32:3435–3446. [PubMed: 21295849]

52. Amoozgar Z, Park J, Lin Q, Yeo Y. Low molecular-weight chitosan as a pH-sensitive


stealth coating for tumor-specific drug delivery. Mol. Pharm. 2012; 9:1262–1270. [PubMed:
22489704]

53. Minchin RF, Martin DJ. Nanoparticles for molecular imaging - an overview.
Endocrinology. 2010; 151:474–481. [PubMed: 20016027]

54. Islam T, Harisinghani MG. Overview of nanoparticle use in cancer imaging. Cancer
Biomark. 2009; 5:61–67. [PubMed: 19414922]

55. Will O, Purkayastha S, Chan C, Athanasiou T, Darzi AW, Gedroyc W, Tekkis PP.
Diagnostic precision of nanoparticle-enhanced MRI for lymph-node metastases: a meta-
analysis. Lancet Oncol. 2006; 7:52–60. [PubMed: 16389184]

56. Peng XH, Qian X, Mao H, Wang AY, Chen ZG, Nie S, Shin DM. Targeted magnetic iron
oxide nanoparticles for tumor imaging and therapy. Int. J. Nanomed. 2008; 3:311–321.

57. Yang L, Peng XH, Wang YA, Wang X, Cao Z, Ni C, Karna P, Zhang X, Wood WC, Gao
X, Nie S, Mao H. Receptor-targeted nanoparticles for in vivo imaging of breast cancer. Clin.
Cancer Res. 2009; 15:4722–4732. [PubMed: 19584158]

58. Hadjipanayis CG, Machaidze R, Kaluzova M, Wang L, Schuette AJ, Chen H, Wu X, Mao
H. EGFRvIII antibody-conjugated iron oxide nanoparticles for magnetic resonance imaging-
guided convection-enhanced delivery and targeted therapy of glioblastoma. Cancer Res.
2010; 70:6303– 6312. [PubMed: 20647323]

59. Xie J, Liu G, Eden HS, Ai H, Chen X. Surface-engineered magnetic nanoparticle


platforms for cancer imaging and therapy. Acc. Chem. Res. 2011; 44:883–892. [PubMed:
21548618]

25
60. Perrault SD, Chan WC. In vivo assembly of nanoparticle components to improve targeted
cancer imaging. Proc. Natl. Acad. Sci. USA. 2010; 107:11194–11199. [PubMed: 20534561]
61. Bogdanov A Jr, Mazzanti ML. Molecular magnetic resonance contrast agents for the
detection of cancer: past and present. Semin. Oncol. 2011; 38:42–54. [PubMed: 21362515]

62. Bogdanov A Jr, Matuszewski L, Bremer C, Petrovsky A, Weissleder R. Oligomerization


of paramagnetic substrates result in signal amplification and can be used for MR imaging of
molecular targets. Mol. Imaging. 2002; 1:16–23. [PubMed: 12920857]

63. Hainfeld JF, Slatkin DN, Focella TM, Smilowitz HM. Gold nanoparticles: a new X-ray
contrast agent. Br. J. Radiol. 2006; 79:248–253. [PubMed: 16498039]

64. Chithrani DB, Dunne M, Stewart J, Allen C, Jaffray DA. Cellular uptake and transport of
gold nanoparticles incorporated in a liposomal carrier. Nanomedicine. 2010; 6:161–169.
[PubMed: 19447206]

65. Chithrani DB, Jelveh S, Jalali F, van Prooijen M, Allen C, Bristow RG, Hill RP, Jaffray
DA. Gold nanoparticles as radiation sensitizers in cancer therapy. Radiat. Res. 2010;
173:719–728. [PubMed: 20518651]

66. Ahmed N, Fessi H, Elaissari A. Theranostic applications of nanoparticles in cancer. Drug


Discov. Today. 2012; 17:928–934. [PubMed: 22484464]

67. Smith L, Kuncic Z, Ostrikov K, Kumar S. Nanoparticles in cancer imaging and therapy.
J. Nanomater. 2012; 2012:1–7.

68. Yoo D, Lee JH, Shin TH, Cheon J. Theranostic magnetic nanoparticles. Acc. Chem. Res.
2011; 44:863–874. [PubMed: 21823593]

69. Xie J, Lee S, Chen X. Nanoparticle-based theranostic agents. Adv. Drug Deliv. Rev.
2010; 62:1064–1079. [PubMed: 20691229]

70. Kohler N, Sun C, Wang J, Zhang M. Methotrexate-modified superparamagnetic


nanoparticles and their intracellular uptake into human cancer cells. Langmuir. 2005;
21:8858–8864. [PubMed: 16142971]

71. Hwu JR, Lin YS, Josephrajan T, Hsu MH, Cheng FY, Yeh CS, Su WC, Shieh DB.
Targeted paclitaxel by conjugation to iron oxide and gold nanoparticles. J. Am. Chem. Soc.
2009; 131:66– 68. [PubMed: 19072111]

26
72. Yu MK, Jeong YY, Park J, Park S, Kim JW, Min JJ, Kim K, Jon S. Drug-loaded
superparamagnetic iron oxide nanoparticles for combined cancer imaging and therapy in
vivo. Angew. Chem. Int. Ed. Engl. 2008; 47:5362–5365. [PubMed: 18551493]

73. Manzoor AA, Lindner LH, Landon CD, Park JY, Simnick AJ, Dreher MR, Das S, Hanna
G, Park W, Chilkoti A, Koning GA, ten Hagen TL, Needham D, Dewhirst MW. Overcoming
limitations in nanoparticle drug delivery: triggered, intravascular release to improve drug
penetration into tumors. Cancer Res. 2012; 72:5566–5575. [PubMed: 22952218]

74. Ghosh S, Dutta S, Gomes E, Carroll D, D’Agostino R Jr, Olson J, Guthold M, Gmeiner
WH. Increased heating efficiency and selective thermal ablation of malignant tissue with
DNA-encased multiwalled carbon nanotubes. ACS Nano. 2009; 3:2667–2673. [PubMed:
19655728]

75. Love SA, Maurer-Jones MA, Thompson JW, Lin YS, Haynes CL. Assessing nanoparticle
toxicity. Annu. Rev. Anal. Chem. (Palo Alto Calif.). 2012; 5:181–205. [PubMed: 22524221]
76. Elias DR, Poloukhtine A, Popik V, Tsourkas A. Effect of ligand density, receptor density,
and nanoparticle size on cell targeting. Nanomedicine. 2013; 9:194–201. [PubMed:
22687896]

77. Maeda H, Nakamura H, Fang J. The EPR effect for macromolecular drug delivery to
solid tumors: improvement of tumor uptake, lowering of systemic toxicity, and distinct tumor
imaging in vivo. Adv. Drug Deliv. Rev. 2013; 65:71–79. [PubMed: 23088862]

78. Liu C, Liu F, Feng L, Li M, Zhang J, Zhang N. The targeted co-delivery of DNA and
doxorubicin to tumor cells via multifunctional PEI-PEG based nanoparticles. Biomaterials.
2013; 34:2547– 2564. [PubMed: 23332321]

79. Stern ST, McNeil SE. Nanotechnology safety concerns revisited. Toxicol. Sci. 2008;
101:4–21. [PubMed: 17602205]

80. Stinchcombe TE. Nanoparticle albumin-bound paclitaxel: a novel Cremphor-EL-free


formulation of paclitaxel. Nanomedicine (Lond.). 2007; 2:415–423. [PubMed: 17716129]

81. Zhang C, Awasthi N, Schwarz MA, Hinz S, Schwarz RE. Superior antitumor activity of
nanoparticle albumin-bound paclitaxel in experimental gastric cancer. PLoS One. 2013;
8:e58037. [PubMed: 23460921]

27
82. Pardee TS, Gomes E, Jennings-Gee J, Caudell D, Gmeiner WH. Unique dual targeting of
thymidylate synthase and topoisomerase1 by FdUMP[10] results in high efficacy against
AML and low toxicity. Blood. 2012; 119:3561–3570. [PubMed: 22362039]

83. Brewer S, Nair-Gill E, Wei B, Chen L, Li X, Riedinger M, Campbell DO, Wiltzius S,


Satyamurthy N, Phelps ME, Radu C, Witte ON, Braun J. Epithelial uptake of [18F]1-(2′-
deoxy-2′- arabinofuranosyl) cytosine indicates intestinal inflammation in mice.
Gastroenterology. 2010; 138:1266–1275. [PubMed: 20080095]

84. Schwarzenberg J, Radu CG, Benz M, Fueger B, Tran AQ, Phelps ME, Witte ON,
Satyamurthy N, Czernin J, Schiepers C. Human biodistribution and radiation dosimetry of
novel PET probes targeting the deoxyribonucleoside salvage pathway. Eur. J. Nucl. Med.
Mol. Imaging. 2011; 38:711–721. [PubMed: 21127859]

85. Boles KS, Schmieder AH, Koch AW, Carano RA, Wu Y, Caruthers SD, Tong RK,
Stawicki S, Hu G, Scott MJ, Zhang H, Reynolds BA, Wickline SA, Lanza GM. MR
angiogenesis imaging with Robo4-vs. alphaVbeta3-targeted nanoparticles in a B16/F10
mouse melanoma model. FASEB J. 2010; 24:4262–4270. [PubMed: 20585027]

86. Winter PM, Caruthers SD, Kassner A, Harris TD, Chinen LK, Allen JS, Lacy EK, Zhang
H, Robertson JD, Wickline SA, Lanza GM. Molecular imaging of angiogenesis in nascent
Vx-2 rabbit tumors using a novel alpha(nu)beta3-targeted nanoparticle and 1.5 tesla magnetic
resonance imaging. Cancer Res. 2003; 63:5838–5843. [PubMed: 14522907]

87. Castro JE, Melo-Cardenas J, Urquiza M, Barajas-Gamboa JS, Pakbaz RS, Kipps TJ. Gene
immunotherapy of chronic lymphocytic leukemia: a phase I study of intranodally injected
adenovirus expressing a chimeric CD154 molecule. Cancer Res. 2012; 72:2937–2948.
[PubMed: 22505652]

88. Bartlett DW, Davis ME. Impact of tumor-specific targeting and dosing schedule on tumor
growth inhibition after intravenous administration of siRNA-containing nanoparticles.
Biotechnol. Bioeng. 2008; 99:975–985. [PubMed: 17929316]

89. Davis ME, Zuckerman JE, Choi CH, Seligson D, Tolcher A, Alabi CA, Yen Y, Heidel
JD, Ribas A. Evidence of RNAi in humans from systemically administered siRNA via
targeted nanoparticles. Nature. 2010; 464:1067–1070. [PubMed: 20305636]

28
90. Heidel JD, Yu Z, Liu JY, Rele SM, Liang Y, Zeidan RK, Kornbrust DJ, Davis ME.
Administration in non-human primates of escalating intravenous doses of targeted
nanoparticles containing ribonucleotide reductase subunit M2 siRNA. Proc. Natl. Acad. Sci.
USA. 2007; 104:5715–5721. [PubMed: 17379663]

91. Schluep T, Hwang J, Cheng J, Heidel JD, Bartlett DW, Hollister B, Davis ME. Preclinical
efficacy of the camptothecin-polymer conjugate IT-101 in multiple cancer models. Clin.
Cancer Res. 2006; 12:1606–1614. [PubMed: 16533788]

92. Ghazani, AA.; McDermott, S.; Pectasides, M.; Sebas, M.; Mino-Kenudson, M.; Lee, H.;
Weissleder, R.; Castro, CM. Comparison of select cancer biomarkers in human circulating
and bulk tumor cells using magnetic nanoparticles and a miniaturized micro-NMR system.
Nanomedicine. NBM 2013;xx:1-9.

93. Niibe Y, Hayakawa K. Oligometastases and oligo-recurrence: the new era of cancer
therapy. Jpn. J. Clin. Oncol. 2010; 40:107–111. [PubMed: 20047860]

94. Veltri A, Guarnieri T, Gazzera C, Busso M, Solitro F, Fora G, Racca P. Long-term


outcome of radiofrequency thermal ablation (RFA) of liver metastases from colorectal cancer
(CRC): size as the leading prognostic factor for survival. Radiol. Med. 2012; 117:1139–1151.
[PubMed: 22430677]

95. Mylona S, Stroumpouli E, Pomoni M, Galani P, Ntai S, Thanos L. Radiofrequency


ablation of liver metastases from cancer of unknown primary site. Diagn. Interv. Radiol.
2009; 15:297–302. [PubMed: 19908183]

96. Vogl TJ, Naguib NN, Nour-Eldin NE, Mack MG, Zangos S, Abskharon JE, Jost A.
Repeated chemoembolization followed by laser-induced thermotherapy for liver metastasis of
breast cancer. AJR Am. J. Roentgenol. 2011; 196:W66–W72. [PubMed: 21178035]

97. Vogl TJ, Naguib NN, Gruber-Rouh T, Koitka K, Lehnert T, Nour-Eldin NE. Microwave
ablation therapy: clinical utility in treatment of pulmonary metastases. Radiology. 2011;
261:643–651. [PubMed: 22012906

98. Lo SS, Moffatt-Bruce SD, Dawson LA, Schwarz RE, Teh BS, Mayr NA, Lu JJ, Grecula
JC, Olencki TE, Timmerman RD. The role of local therapy in the management of lung and
liver oligometastases. Nat. Rev. Clin. Oncol. 2011; 8:405–416. [PubMed: 21606970]

29
99. Zheng Y, Fernando HC. Surgical and nonresectional therapies for pulmonary metastasis.
Surg. Clin. North Am. 2010; 90:1041–1051. [PubMed: 20955882]

100. Mayo SC, Pawlik TM. Thermal ablative therapies for secondary hepatic malignancies.
Cancer J. 2010; 16:111–117. [PubMed: 20404607]

101. Cornelis F, Balageas P, Le Bras Y, Rigou G, Boutault JR, Bouzgarrou M, Grenier N.


Radiologically-guided thermal ablation of renal tumours. Diagn. Interv. Imaging. 2012;
93:246– 261. [PubMed: 22464994]

102. Lorentzen T, Skjoldbye BO, Nolsoe CP. Microwave ablation of liver metastases guided
by contrast-enhanced ultrasound: experience with 125 metastases in 39 patients. Ultraschall
Med. 2011; 32:492–496. [PubMed: 21259183]

103. Vogl TJ, Farshid P, Naguib NN, Zangos S. Thermal ablation therapies in patients with
breast cancer liver metastases: a review. Eur. Radiol. 2013; 23:797–804. [PubMed:
23064713]

104. Munireddy S, Katz S, Somasundar P, Espat NJ. Thermal tumor ablation therapy for
colorectal cancer hepatic metastasis. J. Gastrointest. Oncol. 2012; 3:69–77. [PubMed:
22811871]

105. Mues AC, Mucksavage P, Graversen JA, Landman J. BioGlue surgical adhesive as a
thermal reflector during laparoscopic cryoablation: effect on iceball size and ablation zone
diameter. JSLS. 2012; 16:23–26. [PubMed: 22906325]

106. Kawaguchi M, Ohno J, Irie A, Fukushima T, Yamazaki J, Nakashima N. Dispersion


stability and exothermic properties of DNA-functionalized single-walled carbon nanotubes.
Int. J. Nanomed. 2011; 6:729–736.

107. Kawaguchi M, Yamazaki J, Ohno J, Fukushima T. Preparation and binding study of a


complex made of DNA-treated single-walled carbon nanotubes and antibody for specific
delivery of a “molecular heater” platform. Int. J. Nanomed. 2012; 7:4363–4371.

108. Bozzini G, Colin P, Nevoux P, Villers A, Mordon S, Betrouni N. Focal therapy of


prostate cancer: energies and procedures. Urol. Oncol. 2013; 31:155–167. [PubMed:
22795500]

30
109. Haen SP, Pereira PL, Salih HR, Rammensee HG, Gouttefangeas C. More than just
tumor destruction: immunomodulation by thermal ablation of cancer. Clin. Dev. Immunol.
2011; 2011:160250. [PubMed: 22242035]

110. Bader MJ, Stepp H, Beyer W, Pongratz T, Sroka R, Kriegmair M, Zaak D, Welschof M,
Tilki D, Stief CG, Waidelich R. Photodynamic therapy of bladder cancer – a phase I Study
Using Hexaminolevulinate (HAL). Urol Oncol. 2012

111. Lindenmann J, Matzi V, Neuboeck N, Anegg U, Baumgartner E, Maier A, Smolle J,


SmolleJuettner FM. Individualized, multimodal palliative treatment of inoperable esophageal
cancer: clinical impact of photodynamic therapy resulting in prolonged survival. Lasers Surg.
Med. 2012; 44:189–198. [PubMed: 22334351]

112. Sawa M, Iwata E, Ishikawa K, Gomi F, Nishida K, Terasaki H. Comparison of different


treatment intervals between bevacizumab injection and photodynamic therapy in combined
therapy for agerelated macular degeneration. Jpn. J. Ophthalmol. 2012; 56:470–475.
[PubMed: 22678809]

113. Ethirajan M, Chen Y, Joshi P, Pandey RK. The role of porphyrin chemistry in tumor
imaging and photodynamic therapy. Chem. Soc. Rev. 2010; 40:340–362. [PubMed:
20694259]

114. Chin WW, Heng PW, Thong PS, Bhuvaneswari R, Hirt W, Kuenzel S, Soo KC, Olivo
M. Improved formulation of photosensitizer chlorin e6 polyvinylpyrrolidone for fluorescence
diagnostic imaging and photodynamic therapy of human cancer. Eur. J. Pharm. Biopharm.
2008; 69:1083–1093. [PubMed: 18396019]

115. Rosi NL, Giljohann DA, Thaxton CS, Lytton-Jean AK, Han MS, Mirkin CA.
Oligonucleotidemodified gold nanoparticles for intracellular gene regulation. Science. 2006;
312:1027–1030. [PubMed: 16709779]

116. Wang Z, Liu H, Yang SH, Wang T, Liu C, Cao YC. Nanoparticle-based artificial RNA
silencing machinery for antiviral therapy. Proc. Natl. Acad. Sci. USA. 2012; 109:12387–
12392. [PubMed: 22802676]

117. Fabbro C, Ali-Boucetta H, Da Ros T, Kostarelos K, Bianco A, Prato M. Targeting


carbon nanotubes against cancer. Chem. Commun. (Camb.). 2012; 48:3911–3926. [PubMed:
22428156]

31
118. Gerweck LE. Modification of cell lethality at elevated temperatures. The pH effect.
Radiat. Res. 1977; 70:224–235.

119. Dewey, WF.; Freeman, ML.; Raaphorst, GP. Cellular biology of hyperthermia and
radiation. In: Meyn Raymond, E.; Withers, H. Rodney, editors. Radiation Biology in Cancer
Research. 32nd. New York: Raven Press; 1980. Series: M. D. Anderson Symposia on
Fundamental Cancer Research

120. Szlachcic A, Pala K, Zakrzewska M, Jakimowicz P, Wiedlocha A, Otlewski J. FGF1-


gold nanoparticle conjugates targeting FGFR efficiently decrease cell viability upon NIR
irradiation. Int. J. Nanomed. 2012; 7:5915–5927.

121. You J, Zhang R, Xiong C, Zhong M, Melancon M, Gupta S, Nick AM, Sood AK, Li C.
Effective photothermal chemotherapy using doxorubicin-loaded gold nanospheres that target
EphB4 receptors in tumors. Cancer Res. 2012; 72:4777–4786. [PubMed: 22865457]

122. Silva AC, Oliveira TR, Mamani JB, Malheiros SM, Malavolta L, Pavon LF, Sibov TT,
Amaro E Jr, Tannus A, Vidoto EL, Martins MJ, Santos RS, Gamarra LF. Application of
hyperthermia induced by superparamagnetic iron oxide nanoparticles in glioma treatment.
Int. J. Nanomed. 2011; 6:591–603.

123. Grull H, Langereis S. Hyperthermia-triggered drug delivery from temperature-sensitive


liposomes using MRI-guided high intensity focused ultrasound. J. Control. Release. 2012;
161:317–327. [PubMed: 22565055]

124. Krishnan KM. Biomedical nanomagnetics: a spin through possibilities in imaging,


diagnostics, and therapy. IEEE Trans. Magn. 2010; 46:2523–2558. [PubMed: 20930943]

125. Buzea C, Pacheco II, Robbie K. Nanomaterials and nanoparticles: sources and toxicity.
Biointerphases. 2007; 2:MR17–MR71. [PubMed: 20419892]

32

You might also like